Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 9.035
Filtrar
Más filtros











Intervalo de año de publicación
1.
Toxicol Appl Pharmacol ; 486: 116942, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38692360

RESUMEN

Organotins have been widely used in various industrial applications. This study investigated the structure-activity relationship as inhibitors of human, pig, and rat gonadal 3ß-hydroxysteroid dehydrogenases (3ß-HSD). Human KGN cell, pig, and rat testis microsomes were utilized to assess the inhibitory effects of 18 organotins on the conversion of pregnenolone to progesterone. Among them, diphenyltin, triethyltin, and triphenyltin exhibited significant inhibitory activity against human 3ß-HSD2 with IC50 values of 114.79, 106.98, and 5.40 µM, respectively. For pig 3ß-HSD, dipropyltin, diphenyltin, triethyltin, tributyltin, and triphenyltin demonstrated inhibitory effects with IC50 values of 172.00, 100.19, 87.00, 5.75, and 1.65 µM, respectively. Similarly, for rat 3ß-HSD1, dipropyltin, diphenyltin, triethyltin, tributyltin, and triphenyltin displayed inhibitory activity with IC50 values of 81.35, 43.56, 55.55, 4.09, and 0.035 µM, respectively. They were mixed inhibitors of pig and rat 3ß-HSD, while triphenyltin was identified as a competitive inhibitor of human 3ß-HSD2. The mechanism underlying the inhibition of organotins on 3ß-HSD was explored, revealing that they may disrupt the enzyme activity by binding to cysteine residues in the catalytic sites. This proposition was supported by the observation that the addition of dithiothreitol reversed the inhibition caused by all organotins except for triethyltin, which was partially reversed. In conclusion, this study provides valuable insights into the structure-activity relationship of organotins as inhibitors of human, pig, and rat gonadal 3ß-HSD. The mechanistic investigation suggests that these compounds likely exert their inhibitory effects through binding to cysteine residues in the catalytic sites.


Asunto(s)
Inhibidores Enzimáticos , Compuestos Orgánicos de Estaño , Testículo , Animales , Humanos , Relación Estructura-Actividad , Compuestos Orgánicos de Estaño/farmacología , Compuestos Orgánicos de Estaño/química , Ratas , Masculino , Testículo/enzimología , Testículo/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Porcinos , 3-Hidroxiesteroide Deshidrogenasas/antagonistas & inhibidores , 3-Hidroxiesteroide Deshidrogenasas/metabolismo , Simulación del Acoplamiento Molecular , Progesterona/farmacología , Progesterona/metabolismo , Microsomas/enzimología , Microsomas/efectos de los fármacos , Ratas Sprague-Dawley
2.
Eur J Med Chem ; 260: 115717, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37598483

RESUMEN

Receptor-interacting protein kinases 2 and 3 (RIPK2 and RIPK3) are considered attractive therapeutic enzyme targets for the treatment of a multitude of inflammatory diseases and cancers. In this study, we developed three interrelated series of novel quinazoline-based derivatives to investigate the effects of extensive modifications of positions 6 and 7 of the central core on the inhibitory activity and the selectivity against these RIPKs. The design of the derivatives was inspired by analyses of available literary knowledge on both RIPK2 and RIPK3 in complex with known quinazoline or quinoline inhibitors. Enzymatic investigations for bioactivity of the prepared molecules against purified RIPKs (RIPK1-4) shed light on multiple potent and selective RIPK2 and dual RIPK2/3 inhibitors. Furthermore, evaluations in living cells against the RIPK2-NOD1/2-mediated signaling pathways, identified as the potential primary targets, demonstrated nanomolar inhibition for a majority of the compounds. In addition, we have demonstrated overall good stability of various lead inhibitors in both human and mouse microsomes and plasma. Several of these compounds also were evaluated for selectivity across 58 human kinases other than RIPKs, exhibiting outstanding specificity profiles. We have thus clearly demonstrated that tuning appropriate substitutions at positions 6 and 7 of the developed quinazoline derivatives may lead to interesting potency and specificities against RIPK2 and RIPK3. This knowledge might therefore be employed for the targeted preparation of new, highly potent and selective tools against these RIPKs, which could be of utility in biological and clinical research.


Asunto(s)
Microsomas , Quinazolinas , Humanos , Animales , Ratones , Quinazolinas/farmacología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor
3.
J Sci Food Agric ; 103(12): 5883-5892, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37115015

RESUMEN

BACKGROUND: Cadmium (Cd), known as a vital contaminant in the environment, penetrates the blood-brain barrier and accumulates in the cerebrum. Acute toxicosis of Cd, which leads to lethal cerebral edema, intracellular accumulation and cellular dysfunction, remains to be illuminated with regard to the exact molecular mechanism of cerebral toxicity. Resveratrol (RES), present in the edible portions of numerous plants, is a simply acquirable and correspondingly less toxic natural compound with neuroprotective potential, which provides some theoretical bases for antagonizing Cd-induced cerebral toxicity. RESULTS: This work was executed to research the protective effects of RES against Cd-induced toxicity in chicken cerebrum. Markedly, these lesions were increased in the Cd group, which also exhibited a thinner cortex, reduced granule cells, vacuolar degeneration, and an enlarged medullary space in the cerebrum. Furthermore, Cd induced CYP450 enzyme metabolism disorders by disrupting the nuclear xenobiotic receptor response (NXRs), enabling the cerebrum to reduce the ability to metabolize exogenous substances, eventually leading to Cd accumulation. Meanwhile, accumulated Cd promoted oxidative damage and synergistically promoted the damage to neurons and glial cells. CONCLUSION: RES initiated NXRs (especially for aromatic receptor and pregnancy alkane X receptor), decreasing the expression of CYP450 genes, changing the content of CYP450, maintaining CYP450 enzyme normal activities, and exerting antagonistic action against the Cd-induced abnormal response of nuclear receptors. These results suggest that the cerebrum toxicity caused by Cd was reduced by pretreatment with RES. © 2023 Society of Chemical Industry.


Asunto(s)
Cadmio , Cerebro , Resveratrol/farmacología , Resveratrol/metabolismo , Cadmio/toxicidad , Cadmio/metabolismo , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/farmacología , Cerebro/metabolismo , Estrés Oxidativo , Microsomas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
4.
Toxicol Lett ; 373: 132-140, 2023 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-36442682

RESUMEN

Aflatoxin B1 (AFB1) is a human procarcinogen known to be activated by cytochrome P450 (CYP) 1A2 and 3A4. In a previous study AFB1 caused chromosomal rearrangement in a yeast strain genetically engineered for stably expressing human CYP1B1. Yet, further verification of the effect of AFB1 in human cells, a potential role of the aryl hydrocarbon receptor (AhR), and CYP1B1-catalyzed AFB1 metabolism remain unidentified. In this study, a human hepatocyte (L-02) line and a human lymphoblastoid (TK6) cell line were genetically engineered for the expression of human CYP1B1, producing L-02-hCYP1B1 and TK6-hCYP1B1, respectively. They were exposed to AFB1 and analyzed for the formation of micronucleus and elevation of γ-H2AX (indicating double-strand DNA breaks); the metabolites formed by CYP1B1 from AFB1 after incubation of AFB1 with human CYP1B1 isoenzyme microsomes were determined by LC-MS. The results showed significantly more potent induction of micronucleus by AFB1 in L-02-hCYP1B1 and TK6-hCYP1B1 than in the parental (L-02 and TK6) cells, and the effects were reduced by (E)- 2,3',4,5'-tetramethoxystilbene, a specific CYP1B1 inhibitor. In the AFB1- CYP1B1 microsomes incubations AFM1, a known stable metabolite of AFB1, was detected. Moreover, in L-02 and TK6 cells, AFB1 apparently increased the protein levels of AhR, ANRT and CYP1B1, and caused the nuclear translocation of AhR and ARNT, the latter effect being blocked by BAY-218 (an inhibitor of AhR). In conclusion, this study indicates that human CYP1B1 is capable of metabolically activating AFB1 through the AhR signaling pathway.


Asunto(s)
Aflatoxina B1 , Receptores de Hidrocarburo de Aril , Humanos , Aflatoxina B1/toxicidad , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Sistema Enzimático del Citocromo P-450/metabolismo , Citocromo P-450 CYP1B1/genética , Citocromo P-450 CYP1B1/metabolismo , Microsomas/metabolismo , Línea Celular
5.
J. Health Biol. Sci. (Online) ; 10(1): 1-6, 01/jan./2022. tab
Artículo en Inglés | LILACS | ID: biblio-1411461

RESUMEN

Objectives: The purpose of this study was to evaluate the mutagenic potential of fluoxetine and fluoxetine-galactomannan. Methods: Chromosomal aberration test and Salmonella typhimurium/microsome mutagenicity assay. Results: The results showed that fluoxetine (250 µg/mL) can cause chromosomal breaks of treated leukocytes and increase the frequency of reversion of the tester strains of S. typhimurium / microsome assay only at the highest concentration (5 mg/mL), while fluoxetine encapsulated in galactomannan did not cause these changes (leukocytes and S. typhimuriums strains). Conclusion: In summary, fluoxetine showed a mutagenic effect detectable only at high concentrations in both eukaryotic and prokaryotic models. Furthermore, the fluoxetine/galactomannan complex, in this first moment, prevented the mutagenicity attributed to fluoxetine, emphasizing that the present encapsulation process can be an alternative in preventing these effects in vitro.


Objetivos: avaliar o potencial mutagênico da fluoxetina e da fluoxetina-galactomanana. Métodos: Teste de aberração cromossômica e ensaio de mutagenicidade de Salmonella typhimurium /microssoma. Resultados: a fluoxetina (250 µg/mL) pode causar quebras cromossômicas de leucócitos tratados e aumentar a frequência de reversão das cepas testadoras de S. typhimurium /microssoma apenas na concentração mais alta (5 mg/mL), enquanto a fluoxetina encapsulada em galactomanano não causou essas alterações (leucócitos e cepas de S. typhimurium). Conclusão: a fluoxetina mostrou um efeito mutagênico detectável apenas em altas concentrações em modelos eucarióticos e procarióticos. Além disso, o complexo fluoxetina/galactomanan, neste primeiro momento, evitou a mutagenicidade atribuída à fluoxetina, ressaltando que o presente processo de encapsulamento pode ser uma alternativa na prevenção desses efeitos in vitro.


Asunto(s)
Fluoxetina , Aberraciones Cromosómicas , Salmonella typhimurium , Rotura Cromosómica , Microsomas , Pruebas de Mutagenicidad
6.
Biol Pharm Bull ; 45(8): 1116-1123, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35908893

RESUMEN

Flavones, which are distributed in a variety of plants and foods in nature, possess significant biological activities, including antitumor and anti-inflammatory effects, and are metabolized into glucuronides by uridine 5'-diphosphate (UDP)-glucuronosyltransferase (UGT) enzymes in humans. In this study, apigenin, acacetin, and genkwanin, flavones having hydroxyl groups at C5, C7, and/or C4'positions were focused on, and the regioselective glucuronidation in human liver and intestinal microsomes was examined. Two glucuronides (namely, AP-7G and AP-4'G for apigenin, AC-5G and AC-7G for acacetin, and GE-5G and GE-4'G for genkwanin) were formed from each flavone by liver and intestinal microsomes, except for only GE-4'G formation from genkwanin by intestinal microsomes. The order of total glucuronidation activities was liver microsomes > intestinal microsomes for apigenin and acacetin, and liver microsomes < intestinal microsomes for genkwanin. The order of CLint values (x-intercept) based on v versus V/[S] plots for apigenin glucuronidation was AP-7G > AP-4'G in liver microsomes and AP-7G < AP-4'G in intestinal microsomes. The order of CLint values was AC-5G < AC-7G for acacetin and GE-5G < GE-4'G genkwanin glucuronidation in both liver and intestinal microsomes. This suggests that the abilities and roles of UGT enzymes in the glucuronidation of apigenin, acacetin, and genkwanin in humans differ depending on the chemical structure of flavones.


Asunto(s)
Apigenina , Flavonas , Microsomas , Glucurónidos/metabolismo , Glucuronosiltransferasa/metabolismo , Humanos , Intestinos/metabolismo , Hígado/metabolismo , Microsomas/metabolismo , Microsomas Hepáticos/metabolismo
7.
Int J Mol Sci ; 23(7)2022 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-35409366

RESUMEN

Bufalin and other cardiac steroids (CS) have been used for centuries for the treatment of congestive heart failure, arrhythmias, and other maladies. However, toxicity and the small therapeutic window of this family of steroids limit their use. Therefore, attempts to synthesize a potent, but less toxic, CS are of major importance. In the present study, two novel bufalin derivatives were synthesized and some of their pharmacological properties were characterized. The reaction of bufalin with Ishikawa's reagent resulted in the production of two novel bufalin derivatives: bufalin 2,3-ene and bufalin 3,4-ene. The compounds were purified with TLC and HPLC and their structure was verified with UV, NMR, and MS analyses. The biological activities of these compounds were evaluated by testing their ability to inhibit the Na+, K+-ATPase activity of the brain microsomal fraction to induce cytotoxic activity against the NCI-60 human tumor cell line panel and non-cancer human cells, and to increase the force of contraction of quail embryonic heart muscle cells in culture. The two steroids exhibited biological activities similar to those of other CS in the tested experimental systems, but with reduced cytotoxicity, advocating their development as drugs for the treatment of heart failure and arrhythmias.


Asunto(s)
Bufanólidos , Ouabaína , Arritmias Cardíacas/tratamiento farmacológico , Bufanólidos/metabolismo , Bufanólidos/farmacología , Humanos , Microsomas/metabolismo , Ouabaína/farmacología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
8.
Biomed Pharmacother ; 148: 112774, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35240529

RESUMEN

Castanopsis tribuloides belongs to the oak species (Fagaceae) and it is commonly distributed in evergreen forests of Bangladesh, India, Myanmar, Nepal, China, and Thailand. Our present study aimed at uncovering the antipyretic potential of methanol extract of C. tribuloides bark (CTB) in the mice models. Baker's yeast pyrexia model was employed to determine the antipyretic potentials of the extract. Besides, molecular docking and dynamics simulation of CTB phenolic compounds were explored to validate the experimental results and gain insight into the possible antipyretic mechanism of action that can lead to the design and discovery of novel drugs against mPGES-1. The results revealed that CTB (400 mg/kg) significantly inhibited (P < 0.001) the elevated body temperature of mice since 0.5 h, which is more prominent than the standard. At dose 200 mg/kg, the bark extract also produced significant (P < 0.05) antipyretic activity since 2 h. HPLC-DAD analysis identified and quantified nine polyphenolic compounds from the extract, including rutin hydrate, (-) epicatechin, caffeic acid, catechin hydrate, catechol, trans-ferulic acid, p-coumaric acid, vanillic acid, and rosmarinic acid. Molecular docking study suggested probable competition of these phenolic compounds with glutathione, an essential cofactor for microsomal prostaglandin E synthase-1 (mPGES-1) activity. Additionally, RMSF, RMSD, Rg, and hydrogen bonds performed during MD simulations revealed that rutin hydrate (rich in CTB) bound to the mPGES-1 active site in a stable manner and thus inactivating mPGES-1. Therefore, it can be concluded that rutin hydrate reduces pyrexia in mice via downregulating PGE2 synthesis by inhibiting mPGES-1 activity.


Asunto(s)
Fagaceae , Fiebre/patología , Microsomas/efectos de los fármacos , Extractos Vegetales/farmacología , Prostaglandina-E Sintasas/efectos de los fármacos , Rutina/farmacología , Animales , Femenino , Masculino , Ratones , Simulación del Acoplamiento Molecular , Corteza de la Planta , Extractos Vegetales/química , Polifenoles/química , Polifenoles/farmacología , Rutina/química
9.
Eur J Med Chem ; 231: 114167, 2022 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-35152061

RESUMEN

Microsomal prostaglandin E2 synthase-1 (mPGES-1) is recognized as a promising therapeutic target for next-generation anti-inflammatory drugs to treat inflammatory diseases. In this study, we report the identification of new, potent and selective inhibitors of human mPGES-1 such as compounds 10, 31 and 49 with IC50 of 0.03-0.09 µM in a cell-free assay of PGE2 production. Compound 10 and 49 also inhibited leukotriene C4 synthase (LTC4S) at sub-µM concentrations (IC50 = 0.7 and 0.4 µM, respectively), affording compounds dually targeting inflammatory PGE2 and cysteinyl leukotriene (cys-LT) biosynthesis. However, compound 31 showed substantial selectivity towards mPGES-1 (IC50 = 0.03 µM) with a decreased inhibitory activity on LTC4S (IC50 = 2.8 µM), and also on other related targets such as FLAP and 5-LO. These oxadiazole thione-benzimidazole derivatives warrant further exploration of new and alternative analogs that may lead to the identification of novel derivatives with potent anti-inflammatory properties.


Asunto(s)
Leucotrieno C4 , Microsomas , Antiinflamatorios/farmacología , Bencimidazoles/farmacología , Dinoprostona , Humanos , Prostaglandina-E Sintasas
10.
Sci Rep ; 11(1): 21576, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732782

RESUMEN

Many agents targeting the colchicine binding site in tubulin have been developed as potential anticancer agents. However, none has successfully made it to the clinic, due mainly to dose limiting toxicities and the emergence of multi-drug resistance. Chalcones targeting tubulin have been proposed as a safe and effective alternative. We have shown previously that quinolone chalcones target tubulin and maintain potent anti-proliferative activity vis-à-vis colchicine, while also having high tolerability and low toxicity in mouse models of cancer and refractivity to multi-drug resistance mechanisms. To identify the most effective anticancer chalcone compound, we synthesized 17 quinolone-chalcone derivatives based on our previously published CTR-17 and CTR-20, and then carried out a structure-activity relationship study. We identified two compounds, CTR-21 [((E)-8-Methoxy-3-(3-(2-methoxyphenyl)-3-oxoprop-1-enyl) quinolin-2(1H)-one)] and CTR-32 [((E)-3-(3-(2-ethoxyphenyl)-3-oxoprop-1-enyl) quinolin-2(1H)-one)] as potential leads, which contain independent moieties that play a significant role in their enhanced activities. At the nM range, CTR-21 and CTR-32 effectively kill a panel of different cancer cells originated from a variety of different tissues including breast and skin. Both compounds also effectively kill multi-drug resistant cancer cells. Most importantly, CTR-21 and CTR-32 show a high degree of selectivity against cancer cells. In silico, both of them dock near the colchicine-binding site with similar energies. Whereas both CTR-21 and CTR-32 effectively prevents tubulin polymerization, leading to the cell cycle arrest at G2/M, CTR-21 has more favorable metabolic properties. Perhaps not surprisingly, the combination of CTR-21 and ABT-737, a Bcl-2 inhibitor, showed synergistic effect in killing cancer cells, since we previously found the "parental" CTR-20 also exhibited synergism. Taken together, CTR-21 can potentially be a highly effective and relatively safe anticancer drug.


Asunto(s)
Chalconas/química , Diseño de Fármacos/métodos , Quinolonas/química , Relación Estructura-Actividad , Animales , Apoptosis , Línea Celular Tumoral , Chalconas/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Citometría de Flujo , Células HeLa , Humanos , Enlace de Hidrógeno , Leucocitos Mononucleares/metabolismo , Células MCF-7 , Ratones , Microsomas/química , Paclitaxel/farmacología , Quinolonas/farmacología , Tubulina (Proteína)/química , Moduladores de Tubulina/farmacología
11.
Future Oncol ; 17(35): 4925-4946, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34672683

RESUMEN

Aim: Epoxide hydrolase is involved in oxidative defenses and is responsible for the activation of carcinogens. The relationship between EPHX1 polymorphisms (Tyr113His and His139Arg) and overall survival (OS) and lung cancer (LC) risk was investigated. Methods: The study comprised 550 cases and 550 controls. Genotyping and statistical analysis were applied. Results: The variant genotypes of EPHX1 polymorphisms exhibited no association with LC risk. The Tyr113His polymorphism exhibited twofold increased odds of lymph node invasion (p = 0.04). The Tyr/His genotype is a risk factor for smokers. Subjects carrying the combined genotype for His139Arg showed better median survival time (MST) and the heterozygous genotype revealed better MST in the case of small-cell lung cancer (SCLC; 11.30 vs 6.73 months; log-rank test: p = 0.02). The heterozygous genotype (His139Arg) had longer MST in patients receiving cisplatin/carboplatin and irinotecan (11.30 vs 7.23; log-rank test: p = 0.007) Conclusion: The Tyr113His polymorphism is associated with LC risk in smokers and is a potential prognostic factor for OS in patients with SCLC after irinotecan.


Lay abstract Microsomal epoxide hydrolase (mEH) is an enzyme that plays a defensive role against chemicals. In this study, the relationship between the variation of the epoxide hydrolase and the risk of lung cancer was investigated and its role in the survival of patients with lung cancer was evaluated. The study comprised 550 cases and 550 controls. Genotyping was carried out using molecular biology tools and was followed by statistical analysis. The variant genotype of the EPHX1 gene was not associated with the risk of lung cancer, even based on histology. The variant form of the EPHX1 gene was found to be a risk factor for smokers. The Tyr/His genotype was associated with the risk of lung cancer in male subjects. Patients carrying the variant form of the EPHX1 gene (His139Arg) experienced better survival. The heterozygous genotype of the EPHX1Tyr113His gene was related to longer survival time in patients who received cisplatin/carboplatin along with irinotecan. The EPHX1 Tyr113His polymorphism is associated with LC risk in smokers and is a potential prognostic factor for OS of patients with small-cell lung cancer (SCLC) after irinotecan therapy and might increase the likelihood of lymph node metastasis; EPHX1His139Arg exhibited better survival, especially in patients with SCLC.


Asunto(s)
Susceptibilidad a Enfermedades , Epóxido Hidrolasas/genética , Epóxido Hidrolasas/metabolismo , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/mortalidad , Microsomas/enzimología , Polimorfismo Genético , Fumar Tabaco/efectos adversos , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Sustitución de Aminoácidos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Genotipo , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Platino (Metal)/administración & dosificación , Pronóstico
12.
PLoS One ; 16(7): e0254315, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34242345

RESUMEN

Aggressiveness of cancers, like prostate cancer, has been found to be associated with elevated expression of the microsomal prostaglandin E synthase-1 (mPGES-1). Here, we investigated whether KH176m (the active metabolite of sonlicromanol), a recently discovered selective mPGES-1 inhibitor, could affect prostate cancer cells-derived spheroid growth. We demonstrated that KH176m suppressed mPGES-1 expression and growth of DU145 (high mPGES-1 expression)-derived spheroids, while it had no effect on the LNCaP cell line, which has low mPGES-1 expression. By addition of exogenous PGE2, we found that the effect of KH176m on mPGES-1 expression and spheroid growth is due to the inhibition of a PGE2-driven positive feedback control-loop of mPGES-1 transcriptional regulation. Cancer stem cells (CSCs) are a subset of cancer cells exhibiting the ability of self-renewal, plasticity, and initiating and maintaining tumor growth. Our data shows that mPGES-1 is specifically expressed in this CSCs subpopulation (CD44+CD24-). KH176m inhibited the expression of mPGES-1 and reduced the growth of spheroids derived from the CSC. Based on the results obtained we propose selective mPGES-1 targeting by the sonlicromanol metabolite KH176m as a potential novel treatment approach for cancer patients with high mPGES-1 expression.


Asunto(s)
Dinoprostona , Prostaglandina-E Sintasas , Neoplasias de la Próstata , Línea Celular Tumoral , Humanos , Oxidorreductasas Intramoleculares , Masculino , Microsomas
13.
J Toxicol Environ Health A ; 84(20): 821-835, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34187333

RESUMEN

The aim of this study was to determine pharmacokinetics of α-amanitin, a toxic bicyclic octapeptide isolated from the poisonous mushrooms, following intravenous (iv) or oral (po) administration in mice using a newly developed and validated liquid chromatography-high resolution mass spectrometry. The iv injected α-amanitin disappeared rapidly from the plasma with high a clearance rate (26.9-30.4 ml/min/kg) at 0.1, 0.2, or 0.4 mg/kg doses, which was consistent with a rapid and a major excretion of α-amanitin via the renal route (32.6%). After the po administration of α-amanitin at doses of 2, 5, or 10 mg/kg to mice, the absolute bioavailability of α-amanitin was 3.5-4.8%. Due to this low bioavailability, 72.5% of the po administered α-amanitin was recovered from the feces. When α-amanitin is administered po, the tissue to plasma area under the curve ratio was higher in stomach > large intestine > small intestine > lung ~ kidneys > liver but not detected in brain, heart, and spleen. The high distribution of α-amanitin to intestine, kidneys, and liver is in agreement with the previously reported major intoxicated organs following acute α-amanitin exposure. In addition, α-amanitin weakly or negligibly inhibited cytochrome P450 and 5'-diphospho-glucuronosyltransferase enzymes activity in human liver microsomes as well as major drug transport functions in mammalian cells overexpressing transporters. Data suggested remote drug interaction potential may be associated with α-amanitin exposure.


Asunto(s)
Alfa-Amanitina/farmacocinética , Venenos/farmacocinética , Animales , Cromatografía Liquida , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Humanos , Hígado/enzimología , Masculino , Espectrometría de Masas , Ratones , Ratones Endogámicos ICR , Microsomas/metabolismo
14.
J Med Chem ; 64(8): 5123-5136, 2021 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-33793232

RESUMEN

The retinoid X receptors (RXR) are ligand-activated transcription factors involved in multiple regulatory networks as universal heterodimer partners for nuclear receptors. Despite their high therapeutic potential in many pathologies, targeting of RXR has only been exploited in cancer treatment as the currently available RXR agonists suffer from exceptional lipophilicity, poor pharmacokinetics (PK), and adverse effects. Aiming to overcome the limitations and to provide improved RXR ligands, we developed a new potent RXR ligand chemotype based on the nonsteroidal anti-inflammatory drug oxaprozin. Systematic structure-activity relationship analysis enabled structural optimization toward low nanomolar potency similar to the well-established rexinoids. Cocrystal structures of the most active derivatives demonstrated orthosteric binding, and in vivo profiling revealed superior PK properties compared to current RXR agonists. The optimized compounds were highly selective for RXR activation and induced RXR-regulated gene expression in native cellular and in vivo settings suggesting them as excellent chemical tools to further explore the therapeutic potential of RXR.


Asunto(s)
Oxaprozina/análogos & derivados , Receptores X Retinoide/agonistas , Animales , Sitios de Unión , Supervivencia Celular/efectos de los fármacos , Cristalografía por Rayos X , Semivida , Humanos , Ligandos , Ratones , Microsomas/metabolismo , Simulación de Dinámica Molecular , Oxaprozina/metabolismo , Oxaprozina/farmacología , Isoformas de Proteínas/agonistas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Pirazoles/química , Pirazoles/metabolismo , Pirazoles/farmacología , Ratas , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Relación Estructura-Actividad
15.
Bioorg Med Chem Lett ; 36: 127825, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33508464

RESUMEN

We analyzed the influence of calculated physicochemical properties of more than 20,000 compounds on their P-gp and BCRP mediated efflux, microsomal stability, hERG inhibition, and plasma protein binding. Our goal was to provide guidance for designing compounds with desired pharmacokinetic profiles. Our analysis showed that compounds with ClogP less than 3 and molecular weight less than 400 will have high microsomal stability and low plasma protein binding. Compounds with logD less than 2.2 and/or basic pKa larger than 5.3 are likely to be BCRP substrates and compounds with basic pKa less than 5.2 and/or acidic pKa less than 13.4 are less likely to inhibit hERG. Based on these results, compounds with MW < 400, ClogP < 3, basic pKa < 5.2 and acidic pKa < 13.4 are likely to have good bioavailability and low hERG inhibition.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas Sanguíneas/metabolismo , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Preparaciones Farmacéuticas/química , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Animales , Proteínas Sanguíneas/química , Química Física , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Ratones , Microsomas/química , Microsomas/metabolismo , Estructura Molecular , Peso Molecular , Proteínas de Neoplasias/química , Ratas , Relación Estructura-Actividad
16.
Biochem J ; 478(3): 619-632, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33427868

RESUMEN

Sulfur-containing amino acid residues function in antioxidative responses, which can be induced by the reactive oxygen species generated by excessive copper and hydrogen peroxide. In all Na+/K+, Ca2+, and H+ pumping P-type ATPases, a cysteine residue is present two residues upstream of the essential aspartate residue, which is obligatorily phosphorylated in each catalytic cycle. Despite its conservation, the function of this cysteine residue was hitherto unknown. In this study, we analyzed the function of the corresponding cysteine residue (Cys-327) in the autoinhibited plasma membrane H+-ATPase isoform 2 (AHA2) from Arabidopsis thaliana by mutagenesis and heterologous expression in a yeast host. Enzyme kinetics of alanine, serine, and leucine substitutions were identical with those of the wild-type pump but the sensitivity of the mutant pumps was increased towards copper and hydrogen peroxide. Peptide identification and sequencing by mass spectrometry demonstrated that Cys-327 was prone to oxidation. These data suggest that Cys-327 functions as a protective residue in the plasma membrane H+-ATPase, and possibly in other P-type ATPases as well.


Asunto(s)
Proteínas de Arabidopsis/química , Arabidopsis/enzimología , Cisteína/química , ATPasas de Translocación de Protón/química , Alquilación , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Proteínas de Arabidopsis/antagonistas & inhibidores , Secuencia Conservada , Cobre/metabolismo , Peróxido de Hidrógeno/metabolismo , Yodoacetamida/farmacología , Cinética , Microsomas/metabolismo , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Conformación Proteica , Dominios Proteicos , ATPasas de Translocación de Protón/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
17.
J Med Chem ; 64(3): 1423-1434, 2021 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-33502198

RESUMEN

Despite the recent advances in cancer therapeutics, highly aggressive cancer forms, such as glioblastoma (GBM), still have very low survival rates. The intracellular scaffold protein syntenin, comprising two postsynaptic density protein-95/discs-large/zona occludens-1 (PDZ) domains, has emerged as a novel therapeutic target in highly malignant phenotypes including GBM. Here, we report the development of a novel, highly potent, and metabolically stable peptide inhibitor of syntenin, KSL-128114, which binds the PDZ1 domain of syntenin with nanomolar affinity. KSL-128114 is resistant toward degradation in human plasma and mouse hepatic microsomes and displays a global PDZ domain selectivity for syntenin. An X-ray crystal structure reveals that KSL-128114 interacts with syntenin PDZ1 in an extended noncanonical binding mode. Treatment with KSL-128114 shows an inhibitory effect on primary GBM cell viability and significantly extends survival time in a patient-derived xenograft mouse model. Thus, KSL-128114 is a novel promising candidate with therapeutic potential for highly aggressive tumors, such as GBM.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Péptidos/química , Péptidos/farmacología , Sinteninas/efectos de los fármacos , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Ensayos Analíticos de Alto Rendimiento , Humanos , Ligandos , Ratones , Microsomas/metabolismo , Modelos Moleculares , Mutación , Unión Proteica , Difracción de Rayos X , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Fundam Clin Pharmacol ; 35(2): 432-434, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33369768

RESUMEN

Remdesivir was recently approved to treat COVID-19. While this antiviral agent delivers clinical benefits, several safety concerns in many cases have been raised. This study reports that remdesivir at nanomolar concentrations inhibits carboxylesterase-2 (CES2) through covalent modifications. CES2 is a major drug-metabolizing enzyme. The combination of high potency with irreversible inhibition concludes that cautions must be exercised when remdesivir is used along with drugs hydrolyzed by CES2.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , Carboxilesterasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Adenosina Monofosfato/efectos adversos , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/efectos adversos , Alanina/farmacología , Alanina/uso terapéutico , Antivirales/efectos adversos , Antivirales/uso terapéutico , Carboxilesterasa/metabolismo , Interacciones Farmacológicas , Humanos , Microsomas/metabolismo , Preparaciones Farmacéuticas/metabolismo , Tenofovir/metabolismo , Tratamiento Farmacológico de COVID-19
19.
Int J Mol Sci ; 21(24)2020 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-33333739

RESUMEN

Butylated hydroxytoluene (BHT) and its analogs, butylated hydroxyanisole (BHA) and tert-butyl-hydroquinone (TBHQ), are widely used synthetic preservatives to inhibit lipid oxidation in the food, cosmetic and pharmaceutical industries. Despite their widespread use, little is known about their human exposure and related biotransformation products. The metabolism of these compounds was investigated using in vitro incubations with human and rat liver fractions. Liquid chromatography coupled to high-resolution tandem mass spectrometry was employed to detect and characterize stable and reactive species formed via oxidative metabolism, as well as phase II conjugates. Several oxidative metabolites have been detected, as well as glutathione, glucuronide, and sulfate conjugates, many of which were not previously reported. A combination of accurate mass measurements, MS/MS fragmentation behavior, and isotope-labeling studies were used to elucidate metabolite structures.


Asunto(s)
Antioxidantes/análisis , Hidroxianisol Butilado/metabolismo , Hidroxitolueno Butilado/metabolismo , Animales , Antioxidantes/química , Hidroxianisol Butilado/análogos & derivados , Hidroxianisol Butilado/química , Hidroxitolueno Butilado/análogos & derivados , Hidroxitolueno Butilado/química , Cromatografía Liquida , Glucurónidos/análisis , Glutatión/análisis , Humanos , Técnicas In Vitro , Microsomas/metabolismo , Fenoles/análisis , Fenoles/metabolismo , Ratas , Sulfatos/análisis , Espectrometría de Masas en Tándem
20.
J Med Chem ; 63(24): 15821-15851, 2020 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-33290061

RESUMEN

Acid ceramidase (AC) is a cysteine hydrolase that plays a crucial role in the metabolism of lysosomal ceramides, important members of the sphingolipid family, a diversified class of bioactive molecules that mediate many biological processes ranging from cell structural integrity, signaling, and cell proliferation to cell death. In the effort to expand the structural diversity of the existing collection of AC inhibitors, a novel class of substituted oxazol-2-one-3-carboxamides were designed and synthesized. Herein, we present the chemical optimization of our initial hits, 2-oxo-4-phenyl-N-(4-phenylbutyl)oxazole-3-carboxamide 8a and 2-oxo-5-phenyl-N-(4-phenylbutyl)oxazole-3-carboxamide 12a, which resulted in the identification of 5-[4-fluoro-2-(1-methyl-4-piperidyl)phenyl]-2-oxo-N-pentyl-oxazole-3-carboxamide 32b as a potent AC inhibitor with optimal physicochemical and metabolic properties, showing target engagement in human neuroblastoma SH-SY5Y cells and a desirable pharmacokinetic profile in mice, following intravenous and oral administration. 32b enriches the arsenal of promising lead compounds that may therefore act as useful pharmacological tools for investigating the potential therapeutic effects of AC inhibition in relevant sphingolipid-mediated disorders.


Asunto(s)
Ceramidasa Ácida/antagonistas & inhibidores , Diseño de Fármacos , Inhibidores Enzimáticos/síntesis química , Oxazolona/química , Ceramidasa Ácida/metabolismo , Administración Oral , Animales , Sitios de Unión , Línea Celular Tumoral , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Semivida , Humanos , Concentración 50 Inhibidora , Cinética , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas/metabolismo , Simulación del Acoplamiento Molecular , Oxazolona/metabolismo , Oxazolona/farmacocinética , Solubilidad , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA